You are on page 1of 9

Review series

Inflammation and insulin resistance


Steven E. Shoelson, Jongsoon Lee, and Allison B. Goldfine

Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.

Over a hundred years ago, high doses of salicylates were shown to lower glucose levels in diabetic patients. This should
have been an important clue to link inflammation to the pathogenesis of type 2 diabetes (T2D), but the antihypergly-
cemic and antiinflammatory effects of salicylates were not connected to the pathogenesis of insulin resistance until
recently. Together with the discovery of an important role for tissue macrophages, these new findings are helping to
reshape thinking about how obesity increases the risk for developing T2D and the metabolic syndrome. The evolv-
ing concept of insulin resistance and T2D as having immunological components and an improving picture of how
inflammation modulates metabolism provide new opportunities for using antiinflammatory strategies to correct the
metabolic consequences of excess adiposity.

Historical perspectives on the link between inflammation and other acute-phase reactants (11–13), the findings similarly
and insulin resistance failed to influence thoughts about pathogenesis. More recently,
Clues to the involvement of inflammation in diabetes date back additional epidemiological studies confirmed and extended these
to more than a century ago, when high doses of sodium salicylate early findings (14). Increased levels of markers and mediators
(5.0–7.5 g/d) were first demonstrated to diminish glycosuria in of inflammation and acute-phase reactants such as fibrinogen, 
diabetic patients having “the milder form of the disease,” presum- C-reactive protein (CRP), IL-6, plasminogen activator inhibi-
ably type 2 diabetes (T2D) (1–3). In 1876 Ebstein concluded that tor-1 (PAI-1), sialic acid, and white cell count correlate with inci-
sodium salicylate could make the symptoms of diabetes mellitus dent T2D (15–25). Markers of inflammation and coagulation are
totally disappear (1, 3). Similarly, in 1901 Williamson found that reduced with intensive lifestyle intervention, as was performed
“sodium salicylate had a definite influence in greatly diminish- in the diabetes prevention program (26), but experiments show-
ing the sugar excretion” (2). The effect was rediscovered in 1957 ing that adipose tissue–derived proinflammatory cytokines such
when an insulin-treated diabetic, given high-dose aspirin to treat as TNF-α could actually cause insulin resistance in experimental
the arthritis associated with rheumatic fever, no longer required models provided the necessary impetus to begin thinking in terms
daily insulin injections (4). Fasting and postchallenge glucose of pathogenesis (27–29). This discovery gave the field a critical
concentrations were nearly normal, despite the discontinuation boost, because epidemiological studies, while highly informative,
of insulin and treatment with aspirin alone. Upon resolution of are correlative by nature and, alone, are unable to determine cau-
the joint symptoms, the aspirin was discontinued, and a repeat sality. These different areas of research have coalesced sufficiently
glucose tolerance test was grossly abnormal. Intrigued by these that credible hypotheses can now link inflammation to the devel-
findings, Reid and colleagues prospectively studied 7 additional opment of insulin resistance and pathogenesis of T2D (30, 31).
patients, 4 the “overweight mild type” and three “lean more severe
diabetics” (4). Over a 2-week course of high-dose aspirin (5.0–8.0 Molecular pathways that link inflammation
g/d), fasting blood glucose levels fell from an average of more than and insulin resistance
190 mg/dl before treatment to 92 mg/dl; every patient responded. Hotamisligil and colleagues (27) and Karasik and colleagues (28)
Additional trials showed equivalent efficacy (5, 6). Mechanistic first showed that the proinflammatory cytokine TNF-α was able
studies focused on insulin secretion, undoubtedly because of the to induce insulin resistance. This was a revolutionary idea, that
established importance of insulin secretion in the pathogenesis a substance produced by fat — and overproduced by expanded
of diabetes, but the findings were inconclusive (7). Insulin resis- fat — had local and potentially systemic effects on metabolism.
tance and its role in the pathogenesis of T2D were less well appre- The concept of fat as a site for the production of cytokines and
ciated, and, as a result, insulin sensitization was not considered as other bioactive substances quickly extended beyond TNF-α to
a potential mechanism for glucose lowering at the time. It wasn’t include leptin, IL-6, resistin, monocyte chemoattractant protein-1 
until much later that studies looking at a role for inflammation in (MCP-1), PAI-1, angiotensinogen, visfatin, retinol-binding pro-
the pathogenesis of insulin resistance reinvestigated the hypogly- tein-4, serum amyloid A (SAA), and others (32–36). Adiponectin is
cemic actions of salicylates and identified the molecular target to similarly produced by fat, but expression decreases with increased
be the IκB kinase-β (IKKβ)/NF-κB axis (8–10). adiposity (37). While leptin and adiponectin are true adipokines
Although epidemiological associations relating inflammation that appear to be produced exclusively by adipocytes, TNF-α, IL-6, 
to T2D or obesity can be traced to the late 1950s and 1960s, when MCP-1, visfatin, and PAI-1 are expressed as well at high levels in
increases were found in circulating concentrations of fibrinogen activated macrophages and/or other cells. The relative amount of
each produced by the adipocyte versus associated adipose tissue
Nonstandard abbreviations used: AGE, advanced glycation end product; CRP, macrophages remains unknown. Sites of resistin production are
C-reactive protein; CVD, cardiovascular disease; IKKβ, IκB kinase-β; IRS-1, insulin
more complex; they include macrophages in humans but both
receptor substrate-1; MCP-1, monocyte chemoattractant protein-1; MIP, macrophage
inflammatory protein; PAI-1, plasminogen activator inhibitor-1; RAGE, receptor for adipocytes and macrophages in rodents (34). TNF-α, IL-6, resis-
advanced glycation end products; T2D, type 2 diabetes; TZD, thiazolidinedione. tin, and undoubtedly other pro- or antiinflammatory cytokines
Conflict of interest: The authors have declared that no conflict of interest exists. appear to participate in the induction and maintenance of the
Citation for this article: J. Clin. Invest. 116:1793–1801 (2006). doi:10.1172/JCI29069. subacute inflammatory state associated with obesity. MCP-1

The Journal of Clinical Investigation      http://www.jci.org      Volume 116      Number 7      July 2006 1793
review series

Figure 1
Potential cellular mechanisms for activating
inflammatory signaling. Obesity and high-fat
diet activate IKKβ/NF-κB and JNK pathways
in adipocytes, hepatocytes, and associated
macrophages. Stimuli that have been shown to
activate these pathways during metabolic dys-
regulation include ligands for TNF-α, IL-1, Toll,
or AGE receptors (TNFR, IL-1R, TLR, or RAGE,
respectively), intracellular stresses including
ROS and ER stress, ceramide, and various PKC
isoforms. Obesity-induced IKKβ activation leads
to NF-κB translocation and the increased expres-
sion of numerous markers and potential media-
tors of inflammation that can cause insulin resis-
tance. Obesity-induced JNK activation promotes
the phosphorylation of IRS-1 at serine sites that
negatively regulate normal signaling through the
insulin receptor/IRS-1 axis. Examples include
serine-302 (pS302) and serine-307 (pS307). By
contrast, evidence has not been reported for obe-
sity-induced effects on transcription factors such
as AP-1 that are regulated by JNK. IKKβ and/or
NF-κB are inhibited or repressed by the actions
of salicylates, TZDs, and statins.

and other chemokines have essential roles in the recruitment of face proteins that recognize foreign substances. These include the
macrophages to adipose tissue. These cytokines and chemokines TLRs and the receptor for advanced glycation end products (RAGE)
activate intracellular pathways that promote the development of (44). Many TLR ligands are microbial products, including LPS and
insulin resistance and T2D. lipopeptides derived from bacteria (44). The fact that TLRs recog-
The investigations that focused on intracellular pathways activat- nize microbial lipid conjugates has led to speculation that endog-
ed by inflammation, instead of individual cytokines, have helped enous lipids or lipid conjugates might also activate 1 or more of the
to restructure the framework for thinking about insulin resistance. TLRs in obesity, a possibility supported by experiments showing
As mentioned above, the antihyperglycemic effects of salicylates that saturated fatty acids bind and activate TLR4 (45). Likewise,
focused attention on IKKβ and NF-κB (38–40). However, increasing RAGE binds a variety of ligands, including endogenous advanced
adiposity activates both JNK and IKKβ (8, 41–43). Many of the more glycation end products (AGEs) and a distinct set of microbial prod-
typical proinflammatory stimuli simultaneously activate JNK and ucts (46, 47). AGEs are nonenzymatic adducts formed between
IKKβ pathways, including cytokines and TLRs (Figure 1). Concor- glucose and targeted proteins, particularly those with slow rates
dantly, genetic or chemical inhibition of either JNK or IKKβ/NF-κB  of turnover. Prolonged hyperglycemia and the accompanying pro-
can improve insulin resistance. The several hypothesized mecha- duction of excess quantities of AGEs can activate NF-κB.
nisms that might explain how obesity activates JNK and NF-κB can In addition to proinflammatory cytokine and pattern recogni-
be separated into receptor and nonreceptor pathways (Figure 1). tion receptors, cellular stresses activate JNK and NF-κB, including
Proinflammatory cytokines such as TNF-α and IL-1β activate JNK ROS and ER stress. Systemic markers of oxidative stress increase
and IKKβ/NF-κB through classical receptor-mediated mechanisms with adiposity, consistent with a role for ROS in the development
that have been well characterized (Figure 1). JNK and IKKβ/NF-κB of obesity-induced insulin resistance (48). One potential mecha-
are also activated by pattern recognition receptors, defined as sur- nism is through the activation of NADPH oxidase by lipid accu-

1794 The Journal of Clinical Investigation      http://www.jci.org      Volume 116      Number 7      July 2006
review series

that they do so through dis-


Box 1 similar mechanisms. JNK is
NF-κB target gene products with potential involvement in the pathogenesis of insulin resistance, a stress kinase that normally
T2D, and/or atherosclerosis phosphorylates the c-Jun com-
ponent of the AP-1 transcrip-
Cytokines and Transcription Receptors and Others
tion factor, but to date there are
chemokines factors surface proteins
no known links between this
TNF-α TNFR p55 p65 RelA PAI-1 well-established transcription-
IFN-γ TNFR p75 NF-κB p50 SAA
al pathway and JNK-induced
Resistin IFNR subunits IKKa Angiotensinogen
IL-1β IL-1R IKKβ CRP insulin resistance. Instead, JNK
IL-6 IL-6R IκBα COX2 has been shown to promote
IL-8 CD40 A20 iNOS insulin resistance through the
IL-10 CD40 ligand VEGF phosphorylation of serine resi-
IL-12 E-selectin IGFBPs dues in IRS-1 (41, 42, 51, 57,
IL-18, P-selectin MnSOD 58) (Figure 1). Insulin receptor
Lymphotoxins ICAM-1
signaling that normally occurs
TGF-β VCAM-1
MCP-1 CCR2 through a tyrosine kinase cas-
MIP-1α TLR2 cade is inhibited by counter-
MIP-1β TLR4 regulatory serine/threonine
MIP-2 Lox-1 phosphorylations (59).
MIP-3α RAGE In contrast, IKKβ is highly
RANTES selective toward its physiologi-
IFNR, IFN receptor; IGFBP, IGF-binding protein; IL-1R, IL-1 receptor; MnSOD, manganese superoxide dismutase; SAA,
cal substrates, the IκB protein
serum amyloid A; TNFR, TNF receptor. inhibitors of NF-κB. Phos-
phorylation by IKKβ targets
IκBα for proteasomal degrada-
tion, which liberates NF-κB for
mulation in the adipocyte, which increases ROS production (49). translocation into the nucleus, where it promotes the expression
This mechanism was shown to increase the production of TNF-α,  of numerous target genes whose products induce insulin resis-
IL-6, and MCP-1, and decrease the production of adiponectin. tance (see Box 1). Unlike JNK, IKKβ does not phosphorylate IRS-1
Consistent with this, the antioxidant N-acetyl cysteine can reduce to cause insulin resistance but causes insulin resistance through
ROS and improve insulin resistance in a hyperglycemia-induced transcriptional activation of NF-κB. Increased lipid deposition in
model (50). Lipid accumulation also activates the unfolded pro- adipocytes leads to the production of proinflammatory cytokines,
tein response to increase ER stress in fat and liver (51). ER stress including TNF-α, IL-6, IL-1β, and resistin, which further activate
was shown to activate JNK to lead to serine phosphorylation of JNK and NF-κB pathways through a feed-forward mechanism. In
insulin receptor substrate-1 (IRS-1), but as with all of the stimuli addition to the cytokines, there is upregulated expression of tran-
described in Figure 1, ER stress similarly activates NF-κB. scriptions factors, receptors, and other relevant proteins including
Ceramides may form under conditions of cell stress and promote chemokines that recruit monocytes and stimulate their differen-
cellular signaling, including the regulation of apoptosis. Saturated tiation into macrophages (Box 1).
fats may also promote the synthesis of ceramides, which accumu- Inflammation is also closely linked to the pathogenesis of
late in tissues such as muscle and may correlate with the degree of atherosclerosis, suggesting that inflammation might be a com-
insulin resistance (52). Ceramides can also activate inflammatory mon denominator that links obesity to many of its pathological
pathways including JNK and NF-κB (53). Lipid excess increases the sequelae. Overlapping collections of transcriptionally regulated
activities of various PKC isoforms. Acute lipid infusion activates inflammatory proteins participate in the pathogenesis of these
PKC-θ in rodent muscle and PKC-βII and PKC-δ in human muscle, disorders (Box 1). Signs of inflammation accompany even the
which may also activate IKKβ and NF-κB (54, 55). Consistent with earliest accumulation of lipid within the arterial wall, including
these findings, salicylates improve muscle insulin resistance after the upregulation of the cell adhesion molecules P- and E-selec-
lipid infusion (56), although the relevance of the acute lipid infu- tin, ICAM-1, and VCAM-1, which localizes circulating immune
sion model to the pathology of obesity-induced insulin resistance is cells (60, 61). The adherent cells can migrate into the subendo-
questionable. One or more of the upstream activators of inflamma- thelial space, where they contribute to the local inflammatory
tion listed in Figure 1 may be relevant to the in vivo pathogenesis response. Cytokines and chemokines produced locally include
of insulin resistance, but more information is needed to determine MCP-1 and macrophage inflammatory protein-1α (MIP-1α),
which of these or others predominate. Once activated, however, the MIP-1β, MIP-2, and MIP-3α. T cell activation leads to expres-
processes can be self-perpetuating through a positive-feedback loop sion of IFN-γ and lymphotoxin; macrophages, endothelial cells,
created by the produced proinflammatory cytokines. and SMCs produce TNF-α (62, 63); and together these stimulate
the local production of IL-6 in the atheroma (64, 65). The addi-
Transcription versus phosphorylation tional engagement of CD40 and CD40 ligand promotes these
in the pathogenesis of insulin resistance processes as well as increasing MMP expression (66). The latter
While both JNK and IKKβ/NF-κB play important roles in inflam- can break down collagen and tissue factor, an important media-
mation-induced insulin resistance, accumulated evidence suggests tor of thrombosis (67).

The Journal of Clinical Investigation      http://www.jci.org      Volume 116      Number 7      July 2006 1795
review series

Figure 2
Potential mechanisms for activation of inflammation in adipose tissue. Dietary excess and obesity cause lipid accumulation in adipocytes, initi-
ating a state of cellular stress and activation of JNK and NF-κB. These inflammatory signaling pathways regulate protein phosphorylation and
cellular transcriptional events, thereby leading to increased adipocyte production of proinflammatory cytokines, including TNF-α, IL-6, leptin, and
resistin, chemokines such as MCP-1, and other proatherogenic mediators, for example PAI-1. Endothelial adhesion molecules (e.g., ICAM-1
and VCAM-1) and chemoattractant molecules (designated CCX) bind integrins and chemokine receptors (CCR), respectively, on the monocyte
surface to recruit them to the adipose tissues. The monocytes that differentiate into macrophages produce many of the same inflammatory
cytokines and chemokines as those listed above, and additional ones, to further promote local inflammation and propagate the inflammatory
diathesis systemically. pS, phosphoserine.

As vascular remodeling progresses, the accumulation of foam Where is inflammation-induced


cells leads to the formation of a lipid pool, rich in prothrombotic insulin resistance initiated?
tissue factor. PDGF and TGF-β increase the rate of collagen pro- Adipose tissue has attracted a great deal of attention as a pathogen-
duction, which contributes to the formation of a fibrous cap. ic site of obesity-induced insulin resistance, partly because changes
Conversely, IFN-γ halts collagen synthesis by SMCs. Activated in adiposity are easy to see but also because fat produces bioactive
macrophages secrete MMPs, which degrade collagen and render proteins that are readily detected and reflect the inflammatory
the fibrous cap weak and prone to rupture (68). Thus a dynamic state of the organ. However, it has also been firmly established
balance is maintained between collagen synthesis and breakdown. that all fat is not equal; adipose tissue in the subcutaneous versus
If proinflammatory forces predominate, the fibrous cap may thin abdominal or visceral depots differs by cell size (69, 70), metabolic
and eventually rupture, with release of prothrombotic lipids into activity, and potential role in insulin resistance (71, 72). Visceral fat
the lumen. This may herald the onset of an acute ischemic event. is more pathogenic. The adipocyte itself is integral to the develop-
NF-κB regulates many of the proteins that mediate the atherogenic ment of obesity-induced inflammation. As discussed above, pro-
process, in common with the pathogenesis of insulin resistance, teins produced by adipocytes that might collaboratively initiate the
suggesting that small increases in obesity-induced inflammation process include TNF-α, IL-6, resistin, leptin, adiponectin, MCP-1,
might promote both processes via common mechanisms. This also PAI-1, and angiotensinogen, but since the recruited immune cells
suggests the corollary that pharmacological decreases in inflam- produce many of the same substances, with the exception of leptin
matory activity might coordinately downregulate the production and adiponectin, it is difficult to pinpoint precise sites of produc-
of a number of proteins involved in the pathogenesis of insulin tion. Myeloid-selective deletion of IKKβ improves obesity-induced
resistance, T2D, and cardiovascular disease (CVD). insulin resistance, underscoring potential roles for both NF-κB

1796 The Journal of Clinical Investigation      http://www.jci.org      Volume 116      Number 7      July 2006
review series

Figure 3
Potential mechanisms for adiposity-induced inflam-
mation in the liver. Healthy liver contains a broad
repertoire of cells that participate in inflammatory
and immune responses, including resident hepatic
macrophages (Kupffer cells), B and T cells, NK and
NKT cells, DCs, liver sinusoidal endothelial cells,
hepatic stellate cells, and hepatocytes. Hepatic
steatosis and obesity are accompanied by the acti-
vation of inflammatory signaling pathways in liver.
Proinflammatory cytokines and FFAs, produced
either by hepatocytes in response to steatosis or
by abdominal fat tissue, may activate Kupffer cells.
Numbers of regulatory NKT cells decrease in par-
allel with the Kupffer cell activation.

and inflammatory cells including macrophages (73), but this has ICAM-1, VCAM-1, E-selectin, or P-selectin in response to increased
not helped to distinguish where the process is initiated. Both cell adiposity. As mentioned earlier, MCP-1 induces the migration of
types, lipid-laden adipocytes and recruited macrophages, seem to blood monocytes into the subendothelial space and augments
participate in the pathogenesis of inflammation-induced insulin differentiation into macrophages. Thus changes are predicted in
resistance (Figure 2). Since the bulk of accumulated lipid is stored adipose tissue endothelial cells in response to altered adiposity. As
in adipocytes, it is generally assumed that the adipocyte initiates macrophages rarely function alone, other types of immune cells
the process and the macrophage serves to amplify the signal. Since are likely to participate in adipose tissue inflammation, although
many of the bioactive proteins involved are NF-κB targets, and  this has not yet been reported. In addition to cytokine and protease
NF-κB activation can be self-sustaining, antiinflammatory thera- release, the actions of macrophages in inflammation include anti-
pies including salicylates may coordinately decrease their expres- gen presentation and T cell activation. Classical inflammation
sion and improve insulin resistance. involves the added presence of neutrophils, DCs, NK cells, mast
It is important to understand how increasing adiposity leads to cells, and various subtypes of T lymphocytes. Potential roles for
the recruitment of immune cells to adipose tissue. MCP-1 (CCL2), these other immune cells in adipose tissue inflammation will
a chemoattractant for monocytes, DCs, and memory T cells, is doubtless be topics for future investigation.
produced by adipocytes in parallel with increasing adiposity (74, In addition to adipose tissue, the liver is affected by obesity
75), suggesting that MCP-1 might play a role in recruitment of (Figure 3). Nonalcoholic fatty liver disease (NAFLD) often accom-
monocytes. Consistent with this, mice lacking CCR2, an important panies abdominal adiposity, and its prevalence is increasing and
receptor for MCP-1, are partly protected from developing high-fat closely parallels the prevalence of the comorbid conditions T2D
diet–induced insulin resistance and exhibit reductions in adipose and hyperlipidemia. The pathological spectrum of NAFLD ranges
tissue macrophage recruitment and inflammatory gene expression from simple steatosis to steatohepatitis, advanced fibrosis, and
(76). The fact that protection is incomplete implies that additional cirrhosis. Inflammation clearly plays a pivotal role in the pro-
chemoattractant ligand-receptor pairs might be involved. It is also gression of this disease process. While inadequate suppression of
interesting that some macrophages found in the adipose tissue of hepatic glucose production, due at least in part to hepatic insulin
obese rodents are large and multinucleated (74, 75, 77). Such mul- resistance, is an established contributor to hyperglycemia in T2D,
tinucleate giant cells are often found at sites of chronic inflamma- the role of inflammation in the pathogenesis of these processes
tion and result from the fusion or engulfment of macrophages by has only recently been explored. Inflammatory gene expression
each other. Macrophages including multinucleate giant cells may increases in liver with increasing adiposity (43). This suggests that
aggregate at sites of adipocyte necrosis (77). hepatocyte lipid accumulation (steatosis) might induce a subacute
Other cell types in adipose tissue may also participate in the inflammatory response in liver that is similar to the adipose tis-
inflammatory process. Vascular cells are an obvious place to look. sue inflammation that follows adipocyte lipid accumulation.
Adipose tissue is highly vascularized, with multiple capillaries in Alternatively, proinflammatory substances in the portal circula-
contact with each adipocyte (78). Moreover, adipose tissue rap- tion, potentially produced in abdominal fat, might initiate hepatic
idly proliferates and expands as nutrient stores increase, possibly inflammation. Regardless, NF-κB is activated in the hepatocyte,
using processes similar to the angiogenesis that supports tumor and cytokines including IL-6, TNF-α, and IL-1β are overproduced
growth (77). In addition to being important for fat expansion, the in fatty liver. The proinflammatory cytokines participate in the
microvasculature undoubtedly plays important roles in adipose development of insulin resistance and activate Kupffer cells,
tissue inflammation. For example, circulating leukocytes do not the resident hepatic macrophages. Unlike adipose tissue, where
adhere to normal endothelium, but after initiation of a high-fat macrophages are relatively sparse basally but increase numerically
Western diet the endothelium expresses cell adhesion molecules with adiposity, the liver is densely populated with Kupffer cells,
that bind leukocytes (79). Adipose tissue endothelial cells may which account for over 5% of total cells. The number of Kupffer
increase the expression of one or more of the adhesion proteins cells does not increase with adiposity, but their activation state

The Journal of Clinical Investigation      http://www.jci.org      Volume 116      Number 7      July 2006 1797
review series

Figure 4
Local, portal, and systemic effects of inflammation in insulin resistance and atherogenesis. Increasing adiposity activates inflammatory responses
in fat and liver, with associated increases in the production of cytokines and chemokines. Immune cells including monocytes and macrophages
are recruited and/or activated, and together these cause local insulin resistance. Portal delivery of abdominal fat–derived cytokines and lipids
contributes to hepatic inflammation and insulin resistance. Proinflammatory and proatherogenic mediators are produced in the adipose tissue
and liver and associated immune cells. This creates a systemic inflammatory diathesis that promotes insulin resistance in skeletal muscle and
other tissues and atherogenesis in the vasculature.

does (43). A wide variety of other immune cells are present in nor- neither muscle-specific ablation of IKKβ nor muscle-specific inhi-
mal liver and may also play roles in inflammation-induced insulin bition of NF-κB improves insulin resistance in obese mice (84, 85).
resistance, including T and B lymphocytes, NK cells, and DCs as It is perhaps more appropriate to think of skeletal muscle as a tar-
well as hepatic stellate cells and liver sinusoidal endothelial cells get of inflammation-induced insulin resistance as opposed to a
(80). NKT cells are enriched in normal mouse liver, and their num- site of initiation (Figure 4).
bers decrease in ob/ob or high-fat diet models of obesity (81, 82).
NKT cells are regulatory lymphocytes, with features of both clas- Future trials to target inflammation
sical T (CD3+) and NK (NK1.1+) cells and characteristic expression While demonstrating that TNF-α could induce insulin resistance
of CD1d, an MHC class I homologue that presents glycolipid anti- (27, 28) was paramount in linking inflammation to the pathogene-
gens to TCRs. Adoptive transfer of NKT cells reportedly improves sis of insulin resistance, studies to evaluate whether blocking TNF-α 
nonalcoholic steatohepatitis and glucose intolerance in ob/ob with mAbs improves insulin sensitivity have so far been inconclu-
mice (83), consistent with a role for decreased NKT numbers in sive (86, 87). This may be due in part to small trial sizes and inad-
the pathogenesis of these disorders. equate statistical power but might also be related to mechanisms of
Skeletal muscle is another major site of insulin resistance in obe- pathogenesis. TNF-α by itself may just not be that important. With
sity and T2D. However, increasing adiposity does not appear to multiple cytokines and chemokines involved, it is also possible that
activate inflammatory cascades in skeletal muscle, as it does in fat different combinations of cytokines and chemokines might be
and liver. Inflammation is activated in muscle by intralipid infu- more or less important in different patients. If this were true, then
sion, but this is distinct from the effects of increasing adiposity. neutralization strategies might need to be individually tailored. It
The lipid infusion model is a research tool used to acutely raise may be more prudent to modulate signaling at convergent sites of
circulating and intratissue fatty acid levels and induce insulin integration, such as JNK or IKKβ/NF-κB, since this would provide
resistance. Intralipid infusion activates PKC-θ and IKKβ in mouse a more general strategy for treatment. Several drugs in current clini-
muscle, and the associated insulin resistance is inhibited by salicy- cal practice have been shown to have antiinflammatory properties
late or IKKβ depletion (54, 56). This is in contrast with high-fat or side effects distinct from their major mechanisms of action,
diet–induced and obesity-induced insulin resistance, neither of including members of the thiazolidinedione (TZD) class of PPARγ
which activates IKKβ/NF-κB in skeletal muscle (84, 85) or leads agonists and members of the statin class of HMG CoA reductase
to an increase in skeletal muscle macrophages (75). Concordantly, inhibitors. Both appear to have important antiinflammatory prop-

1798 The Journal of Clinical Investigation      http://www.jci.org      Volume 116      Number 7      July 2006
review series

erties and potential benefits beyond their primary actions on glu- aggregation effects, which, coupled with gastrointestinal irritation,
cose homeostasis and cholesterol lowering, respectively. are associated with unacceptably high risks of bleeding.
TZDs, which include pioglitazone, rosiglitazone, and trogli- Aspirin is a prototypical NSAID that effectively inhibits the
tazone (the latter is no longer in clinical use), are used to improve COX enzymes COX1 and COX2 through the irreversible modifi-
insulin sensitivity and reduce hyperglycemia in patients with T2D. cation of the enzymes’ active sites. This occurs through a trans-
The TZDs’ primary mode of action is through binding and activat- acetylation reaction; the acetyl group of aspirin migrates to and
ing PPARγ to induce the expression of a number of gene products covalently modifies the enzymes to block their catalytic activities.
with roles in adipocyte differentiation, lipid and glucose uptake, In platelets this occurs even at low doses (80–100 mg/d), because
and fatty acid storage (88). TZDs’ beneficial actions in insulin sen- they lack nuclei and cannot resynthesize COX1, and platelets
sitivity are often attributed to fatty acid sequestration in the adi- are thus inactivated for their lifetimes. Aspirin at a typical dose
pose tissue. This reduces circulation levels of FFAs and keeps the of 650 mg covalently modifies COX1 and COX2 in all tissues,
fatty acids out of other tissues, including muscle and liver, where which similarly inhibits prostaglandin synthesis. NSAIDs other
they can cause insulin resistance. But in addition to adipocytes, than aspirin do not transacetylate the COX enzymes but bind
PPARγ is also expressed in macrophages and other immune cells, with sufficiently high affinity that the enzymes are competitively
hepatocytes, endothelial cells, and VSMCs. The antiinflammatory inhibited. The mechanism of action of high-dose salicylate is dis-
actions of the TZDs appear to be mediated through the transre- tinct, as it lacks an acetyl group and neither covalently modifies
pression of NF-κB and consequent decreases in the expression of COX1 or COX2 nor binds with sufficient affinity to effectively
target genes for cytokine and growth factors, cell proliferation and inhibit the enzymes.
migration, ECM remodeling, and cell cycle progression and dif- The nonacetylated salicylates, delivered as sodium salicylate, sal-
ferentiation. One proposed mechanism involves the TZD-depen- salate, and Trilisate, inhibit NF-κB (38–40). This is presumed to be
dent SUMOylation of PPARγ, which targets it to nuclear receptor through direct inhibition of IKKβ (40). The nonacetylated salicy-
corepressor–histone deacetylase-3 (NCoR-HDAC3) complexes lates do not prolong bleeding times and may thus provide a rela-
on inflammatory gene promoters (such as NF-κB and AP-1). The tively safe and effective means of targeting the subacute inflamma-
binding of SUMOylated PPARγ stabilizes the corepressor complex- tion that underlies the obesity-related syndromes. In fact, clinical
es and prevents their clearance, thus maintaining the transcription trials to test efficacy, tolerability, and durability of salsalate (Disal-
factor complexes in a repressed state (89). It has even been sug- cid) are currently being undertaken in the NIH-funded TINSAL-
gested that TZDs exert antiinflammatory effects through PPARγ- T2D and -CVD trials (Targeting Inflammation with Salsalate in
independent activation of the glucocorticoid receptor (90). The T2D or CVD, respectively). Other antiinflammatory strategies are
clinical benefits of TZDs may in part depend upon antiinflam- also being considered, including the potential use of specific JNK
matory effects that work together with classical mechanisms of or IKKβ inhibitors and compounds that block TNF-α, IL-6, TLR,
glucose and lipid regulation (91) to improve insulin sensitivity, or chemokine signaling or reduce oxidative or ER stress, but these
promote plaque remodeling, and potentially reduce cardiovascular studies are at much earlier stages.
events (92, 93). Several other nuclear receptors, including PPARα,
PPARδ, and liver X receptor (LXR), have also been shown to have Concluding remarks
antiinflammatory properties that may be beneficial for the treat- While the importance of inflammation-induced insulin resistance
ment of metabolic or cardiovascular conditions (94, 95), although is doubtless increasing in parallel with the epidemic of obesity,
significant glucose-lowering effects have not been associated with there are additional unrelated mechanisms associated with insulin
agonists to these transcription factors. resistance. For example, polymorphisms in genes encoding com-
Several of the clinically available statins have been shown to down- ponents of the insulin signaling pathway and the insulin-resistant
regulate transcriptional activities of NF-κB, AP-1, and HIF-1α,  syndromes in patients with lipodystrophy who lack adipose tis-
with coordinate reductions in the expression of prothrombotic and sue are different from the insulin resistance in typical overweight
inflammatory cytokines (95). Randomized clinical trials evaluat- patients with T2D. Whether antiinflammatory strategies are ben-
ing statins have also demonstrated reductions in CRP, multiple eficial in these cases has not been investigated. Similarly unknown
cytokines, and inflammatory markers. Despite these modest anti- are the distinctions between populations, such as those of Asian
inflammatory properties, the statins do not appear to significantly descent who may exhibit the characteristics of adiposity-induced
influence either insulin resistance or glycemia. inflammation while relatively lean.
The antiinflammatory properties of TZDs and statins are side In summary, obesity, T2D, and CVD share a metabolic milieu
effects distinct from their primary modes of action. By contrast, characterized by insulin resistance and chronic subacute inflam-
high-dose salicylates directly target inflammation by inhibiting mation. While drugs that secondarily alter the inflammatory pro-
NF-κB (38–40). The glucose-lowering effects of salicylates, seen cess are undoubtedly of great clinical importance, several lines of
decades ago in patients with diabetes (1–7), are now recognized as evidence suggest it might also be possible to directly target inflam-
well to be due at least in part to NF-κB inhibition (8–10, 43, 73). mation with pharmacological interventions to treat and/or prevent
More recent studies demonstrated that high-dose aspirin (∼7.0 g/d)  insulin resistance and T2D and modulate risk for CVD and other
improved multiple metabolic measures in patients with T2D, metabolic conditions. These approaches may provide clinical ben-
including substantial reductions in fasting and postprandial glu- efits to a large number of persons affected by the obesity epidemic
cose, triglycerides, and FFAs (9). These changes were associated and the related cluster of metabolic disorders.
with reduced hepatic glucose production and improvements in
insulin-stimulated glucose disposal, assessed during euglycemic- Address correspondence to: Steven E. Shoelson, Joslin Diabetes
hyperinsulinemic clamping. However, the therapeutic usefulness of Center, One Joslin Place, Boston, Massachusetts 02215, USA. 
high-dose aspirin is limited by the antithrombotic and anti–platelet E-mail: steven.shoelson@joslin.harvard.edu.

The Journal of Clinical Investigation      http://www.jci.org      Volume 116      Number 7      July 2006 1799
review series

1. Ebstein, W. 1876. Zur therapie des Diabetes mel- national sample of US adults. Am. J. Epidemiol. 42. Hirosumi, J., et al. 2002. A central role for JNK in
litus, insbesondere über die Anwendung des sali- 155:57–64. obesity and insulin resistance. Nature. 420:333–336.
cylsauren Natron bei demselben. Berliner Klinische 23. Prince, R.L., Larkins, R.G., and Alford, F.P. 1981. 43. Cai, D., et al. 2005. Local and systemic insulin resis-
Wochenschrift. 13:337–340. The effect of acetylsalicylic acid on plasma glucose tance resulting from hepatic activation of IKKβ
2. Williamson, R.T. 1901. On the treatment of glycos- and the response of glucose regulatory hormones and NF-κB. Nat. Med. 11:183–190.
uria and diabetes mellitus with sodium salicylate. to intravenous glucose and arginine in insulin 44. Akira, S., Uematsu, S., and Takeuchi, O. 2006.
Br. Med. J. 1:760–762. treated diabetics and normal subjects. Metabolism. Pathogen recognition and innate immunity. Cell.
3. Shoelson, S. 2002. Invited comment on W. Ebstein: 30:293–298. 124:783–801.
on the therapy of diabetes mellitus, in particular 24. Nakanishi, N., Yoshida, H., Matsuo, Y., Suzuki, K., 45. Lee, J.Y., Sohn, K.H., Rhee, S.H., and Hwang, D.
on the application of sodium salicylate. J. Mol. Med. and Tatara, K. 2002. White blood-cell count and 2001. Saturated fatty acids, but not unsaturated
80:618–619. the risk of impaired fasting glucose or type II dia- fatty acids, induce the expression of cyclooxygenase-
4. Reid, J., Macdougall, A.I., and Andrews, M.M. 1957. betes in middle-aged Japanese men. Diabetologia. 2 mediated through Toll-like receptor 4. J. Biol.
On the efficacy of salicylate in treating diabetes 45:42–48. Chem. 276:16683–16689.
mellitus. Br. Med. J. 2:1071–1074. 25. Spranger, J., et al. 2003. Inflammatory cytokines 46. Ramasamy, R., Yan, S.F., and Schmidt, A.M. 2005.
5. Hecht, A., and Goldner, M.G. 1959. Reappraisal of and the risk to develop type 2 diabetes: results of The RAGE axis and endothelial dysfunction:
the hypoglycemic action of acetylsalicylate. Metabo- the prospective population-based European Pro- maladaptive roles in the diabetic vasculature and
lism. 8:418–428. spective Investigation into Cancer and Nutrition beyond. Trends Cardiovasc. Med. 15:237–243.
6. Gilgore, S.G. 1960. The influence of salicylate on (EPIC)-Potsdam Study. Diabetes. 52:812–817. 47. Bierhaus, A., et al. 2005. Understanding RAGE,
hyperglycemia. Diabetes. 9:392–393. 26. Haffner, S., et al. 2005. Intensive lifestyle interven- the receptor for advanced glycation end products. 
7. Baron, S.H. 1982. Salicylates as hypoglycemic tion or metformin on inflammation and coagula- J. Mol. Med. 83:876–886.
agents. Diabetes Care. 5:64–71. tion in participants with impaired glucose toler- 48. Keaney, J.F., Jr., et al. 2003. Obesity and systemic
8. Yuan, M., et al. 2001. Reversal of obesity- and diet- ance. Diabetes. 54:1566–1572. oxidative stress: clinical correlates of oxidative
induced insulin resistance with salicylates or tar- 27. Hotamisligil, G.S., Shargill, N.S., and Spiegelman, stress in the Framingham Study. Arterioscler.
geted disruption of Ikkβ. Science. 293:1673–1677. B.M. 1993. Adipose expression of tumor necrosis Thromb. Vasc. Biol. 23:434–439.
9. Hundal, R.S., et al. 2002. Mechanism by which factor-α: direct role in obesity-linked insulin resis- 49. Furukawa, S., et al. 2004. Increased oxidative
high-dose aspirin improves glucose metabolism tance. Science. 259:87–91. stress in obesity and its impact on metabolic syn-
in type 2 diabetes. J. Clin. Invest. 109:1321–1326. 28. Feinstein, R., Kanety, H., Papa, M.Z., Lunenfeld, B., drome. J. Clin. Invest. 114:1752–1761. doi:10.1172/
doi:10.1172/JCI200214955. and Karasik, A. 1993. Tumor necrosis factor-α sup- JCI200421625.
10. Shoelson, S.E., Lee, J., and Yuan, M. 2003. Inflam- presses insulin-induced tyrosine phosphorylation 50. Lin, Y., et al. 2005. The hyperglycemia-induced
mation and the IKKβ/IκB/NF-κB axis in obesity- of insulin receptor and its substrates. J. Biol. Chem. inflammatory response in adipocytes: the role of
and diet-induced insulin resistance. Int. J. Obes. 268:26055–26058. reactive oxygen species. J. Biol. Chem. 280:4617–4626.
Relat. Metab. Disord. 27(Suppl. 3):S49–S52. 29. Hotamisligil, G.S., and Spiegelman, B.M. 1994. 51. Ozcan, U., et al. 2004. Endoplasmic reticulum
11. Fearnley, G.R., Vincent, C.T., and Chakrabarti, R. Tumor necrosis factor alpha: a key component of stress links obesity, insulin action, and type 2 dia-
1959. Reduction of blood fibrinolytic activity in the obesity-diabetes link. Diabetes. 43:1271–1278. betes. Science. 306:457–461.
diabetes mellitus by insulin. Lancet. 2:1067. 30. Pickup, J.C., and Crook, M.A. 1998. Is type II dia- 52. Straczkowski, M., et al. 2004. Relationship between
12. Ogston, D., and McAndrew, G.M. 1964. Fibrinoly- betes mellitus a disease of the innate immune sys- insulin sensitivity and sphingomyelin signal-
sis in obesity. Lancet. 14:1205–1207. tem? Diabetologia. 41:1241–1248. ing pathway in human skeletal muscle. Diabetes.
13. Grace, C.S., and Goldrick, R.B. 1968. Fibrinolysis 31. Kolb, H., and Mandrup-Poulsen, T. 2005. An 53:1215–1221.
and body build. Interrelationships between blood immune origin of type 2 diabetes? Diabetologia. 53. Summers, S.A. 2006. Ceramides in insulin resis-
fibrinolysis, body composition and parameters of 48:1038–1050. tance and lipotoxicity. Prog. Lipid Res. 45:42–72.
lipid and carbohydrate metabolism. J. Atheroscler. 32. Zhang, Y., Proenca, R., Maffei, M., Barone, M., 54. Griffin, M.E., et al. 1999. Free fatty acid-induced
Res. 8:705–719. Leopold, L., and Friedman, J.M. 1994. Positional insulin resistance is associated with activation of
14. Tataranni, P.A., and Ortega, E. 2005. A burning cloning of the mouse obese gene and its human protein kinase C theta and alterations in the insu-
question: does an adipokine-induced activation of homologue. Nature. 372:425–432. lin signaling cascade. Diabetes. 48:1270–1274.
the immune system mediate the effect of overnu- 33. Fried, S.K., Bunkin, D.A., and Greenberg, A.S. 55. Itani, S.I., Ruderman, N.B., Schmieder, F., and
trition on type 2 diabetes? Diabetes. 54:917–927. 1998. Omental and subcutaneous adipose tissues Boden, G. 2002. Lipid-induced insulin resistance
15. Schmidt, M.I., et al. 1999. Markers of inflamma- of obese subjects release interleukin-6: depot dif- in human muscle is associated with changes in dia-
tion and prediction of diabetes mellitus in adults ference and regulation by glucocorticoid. J. Clin. cylglycerol, protein kinase C, and IkappaB-alpha.
(Atherosclerosis Risk in Communities study): a Endocrinol. Metab. 83:847–850. Diabetes. 51:2005–2011.
cohort study. Lancet. 353:1649–1652. 34. Steppan, C.M., et al. 2001. The hormone resistin 56. Kim, J.K., et al. 2001. Prevention of fat-induced
16. Duncan, B.B., et al. 2003. Low-grade systemic links obesity to diabetes. Nature. 409:307–312. insulin resistance by salicylate. J. Clin. Invest.
inflammation and the development of type 2 dia- 35. Shimomura, I., et al. 1996. Enhanced expression of 108:437–446.
betes: the Atherosclerosis Risk in Communities PAI-1 in visceral fat: possible contributor to vascu- 57. Aguirre, V., et al. 2002. Phosphorylation of Ser307
study. Diabetes. 52:1799–1805. lar disease in obesity. Nat. Med. 2:800–803. in insulin receptor substrate-1 blocks interactions
17. Pradhan, A.D., Manson, J.E., Rifai, N., Buring, J.E., 36. Fukuhara, A., et al. 2005. Visfatin: a protein secret- with the insulin receptor and inhibits insulin
and Ridker, P.M. 2001. C-reactive protein, interleu- ed by visceral fat that mimics the effects of insulin. action. J. Biol. Chem. 277:1531–1537.
kin 6, and risk of developing type 2 diabetes mel- Science. 307:426–430. 58. Werner, E.D., Lee, J., Hansen, L., Yuan, M., and
litus. JAMA. 286:327–334. 37. Scherer, P.E., Williams, S., Fogliano, M., Baldini, Shoelson, S.E. 2004. Insulin resistance due to
18. Barzilay, J.I., et al. 2001. The relation of markers G., and Lodish, H.F. 1995. A novel serum protein phosphorylation of insulin receptor substrate-1 at
of inflammation to the development of glucose similar to C1q, produced exclusively in adipocytes. serine 302. J. Biol. Chem. 279:35298–35305.
disorders in the elderly: the Cardiovascular Health J. Biol. Chem. 270:26746–26749. 59. Zick, Y. 2005. Ser/Thr phosphorylation of IRS pro-
Study. Diabetes. 50:2384–2389. 38. Kopp, E., and Ghosh, S. 1994. Inhibition of NF- teins: a molecular basis for insulin resistance. Sci.
19. Vozarova, B., et al. 2002. High white blood cell kappa B by sodium salicylate and aspirin. Science. STKE. 268:e4.
count is associated with a worsening of insulin 265:956–959. 60. Tedder, T.F., Steeber, D.A., Chen, A., and Engel, P.
sensitivity and predicts the development of type 2 39. Pierce, J.W., Read, M.A., Ding, H., Luscinskas, F.W., 1995. The selectins: vascular adhesion molecules.
diabetes. Diabetes. 51:455–461. and Collins, T. 1996. Salicylates inhibit I kappa FASEB J. 9:866–873.
20. Festa, A., D’Agostino, R., Jr., Tracy, R.P., and Haff- B-alpha phosphorylation, endothelial-leukocyte 61. Adams, D.H., and Shaw, S. 1994. Leucocyte-
ner, S.M. 2002. Elevated levels of acute-phase adhesion molecule expression, and neutrophil endothelial interactions and regulation of leuco-
proteins and plasminogen activator inhibitor-1 transmigration. J. Immunol. 156:3961–3969. cyte migration. Lancet. 343:831–836.
predict the development of type 2 diabetes: the 40. Yin, M.J., Yamamoto, Y., and Gaynor, R.B. 1998. 62. Warner, S.J., and Libby, P. 1989. Human vascu-
insulin resistance atherosclerosis study. Diabetes. The anti-inflammatory agents aspirin and salicy- lar smooth muscle cells. Target for and source of
51:1131–1137. late inhibit the activity of IκB kinase-β. Nature. tumor necrosis factor. J. Immunol. 142:100–109.
21. Freeman, D.J., et al. 2002. C-reactive protein is an 396:77–80. 63. Barath, P., et al. 1990. Detection and localization
independent predictor of risk for the development 41. Aguirre, V., Uchida, T., Yenush, L., Davis, R., and of tumor necrosis factor in human atheroma. Am.
of diabetes in the West of Scotland Coronary Pre- White, M.F. 2000. The c-Jun N-terminal kinase J. Cardiol. 65:297–302.
vention Study. Diabetes. 51:1596–1600. promotes insulin resistance during association 64. Seino, Y., et al. 1994. Interleukin 6 gene transcripts
22. Ford, E.S. 2002. Leukocyte count, erythrocyte with insulin receptor substrate-1 and phosphory- are expressed in human atherosclerotic lesions.
sedimentation rate, and diabetes incidence in a lation of Ser(307). J. Biol. Chem. 275:9047–9054. Cytokine. 6:87–91.

1800 The Journal of Clinical Investigation      http://www.jci.org      Volume 116      Number 7      July 2006
review series

65. Rus, H.G., Vlaicu, R., and Niculescu, F. 1996. Invest. 112:1796–1808. doi:10.1172/JCI200319246. with etanercept in obese patients with type 2 diabe-
Interleukin-6 and interleukin-8 protein and gene 76. Weisberg, S.P., et al. 2006. CCR2 modulates inflam- tes. J. Vasc. Res. 42:517–525.
expression in human arterial atherosclerotic wall. matory and metabolic effects of high-fat feeding.  87. Gonzalez-Gay, M.A., et al. 2006. Anti-tumor necro-
Atherosclerosis. 127:263–271. J. Clin. Invest. 116:115–124. doi:10.1172/JCI24335. sis factor-alpha blockade improves insulin resis-
66. Schonbeck, U., and Libby, P. 2001. CD40 signaling 77. Kolonin, M.G., Saha, P.K., Chan, L., Pasqualini, R., tance in patients with rheumatoid arthritis. Clin.
and plaque instability. Circ. Res. 89:1092–1103. and Arap, W. 2004. Reversal of obesity by targeted Exp. Rheumatol. 24:83–86.
67. Mach, F., Schonbeck, U., Bonnefoy, J.Y., Pober, J.S., ablation of adipose tissue. Nat. Med. 10:625–632. 88. Yki-Jarvinen, H. 2004. Thiazolidinediones. N. Engl.
and Libby, P. 1997. Activation of monocyte/mac- 78. Crandall, D.L., Hausman, G.J., and Kral, J.G. 1997. J. Med. 351:1106–1118.
rophage functions related to acute atheroma com- A review of the microcirculation of adipose tissue: 89. Pascual, G., et al. 2005. A SUMOylation-depen-
plication by ligation of CD40: induction of colla- anatomic, metabolic, and angiogenic perspectives. dent pathway mediates transrepression of inflam-
genase, stromelysin, and tissue factor. Circulation. Microcirculation. 4:211–232. matory response genes by PPAR-gamma. Nature.
96:396–399. 79. Blake, G.J., and Ridker, P.M. 2002. Inflammatory 437:759–763.
68. Libby, P. 2001. Current concepts of the pathogen- bio-markers and cardiovascular risk prediction.  90. Ialenti, A., et al. 2005. Mechanism of the anti-
esis of the acute coronary syndromes. Circulation. J. Intern. Med. 252:283–294. inflammatory effect of thiazolidinediones: rela-
104:365–372. 80. Racanelli, V., and Rehermann, B. 2006. The liver as tionship with the glucocorticoid pathway. Mol.
69. Johnson, P.R., and Hirsch, J. 1972. Cellularity of an immunological organ. Hepatology. 43(Suppl. 1): Pharmacol. 67:1620–1628.
adipose depots in six strains of genetically obese S54–S62. 91. Xiang, A.H., et al. 2006. Effect of pioglitazone on
mice. J. Lipid Res. 13:2–11. 81. Guebre-Xabier, M., et al. 2000. Altered hepatic lym- pancreatic beta-cell function and diabetes risk in
70. Krotkiewski, M., Bjorntorp, P., Sjostrom, L., and phocyte subpopulations in obesity-related murine Hispanic women with prior gestational diabetes.
Smith, U. 1983. Impact of obesity on metabolism fatty livers: potential mechanism for sensitization Diabetes. 55:517–522.
in men and women. Importance of regional adipose to liver damage. Hepatology. 31:633–640. 92. Meisner, F., et al. 2006. Effect of rosiglitazone treat-
tissue distribution. J. Clin. Invest. 72:1150–1162. 82. Li, Z., Soloski, M.J., and Diehl, A.M. 2005. Dietary ment on plaque inflammation and collagen con-
71. Coon, P.J., Rogus, E.M., Drinkwater, D., Muller, factors alter hepatic innate immune system in mice tent in nondiabetic patients: data from a random-
D.C., and Goldberg, A.P. 1992. Role of body fat dis- with nonalcoholic fatty liver disease. Hepatology. ized placebo-controlled trial. Arterioscler. Thromb.
tribution in the decline in insulin sensitivity and 42:880–885. Vasc. Biol. 26:845–850.
glucose tolerance with age. J. Clin. Endocrinol. Metab. 83. Elinav, E., et al. 2006. Adoptive transfer of regula- 93. Dormandy, J.A., et al. 2005. Secondary preven-
75:1125–1132. tory NKT lymphocytes ameliorates non-alcoholic tion of macrovascular events in patients with type
72. Gastaldelli, A., et al. 2002. Metabolic effects of vis- steatohepatitis and glucose intolerance in ob/ob 2 diabetes in the PROactive Study (PROspective
ceral fat accumulation in type 2 diabetes. J. Clin. mice and is associated with intrahepatic CD8 trap- pioglitAzone Clinical Trial In macroVascular
Endocrinol. Metab. 87:5098–5103. ping. J. Pathol. 209:121–128. Events): a randomised controlled trial. Lancet.
73. Arkan, M.C., et al. 2005. IKK-beta links inflamma- 84. Rohl, M., et al. 2004. Conditional disruption of IκB 366:1279–1289.
tion to obesity-induced insulin resistance. Nat. Med. kinase 2 fails to prevent obesity-induced insulin 94. Glass, C.K., and Ogawa, S. 2006. Combinato-
11:191–198. resistance. J. Clin. Invest. 113:474–481. doi:10.1172/ rial roles of nuclear receptors in inflammation and
74. Sartipy, P., and Loskutoff, D.J. 2003. Monocyte JCI200418712. immunity. Nat. Rev. Immunol. 6:44–55.
chemoattractant protein 1 in obesity and insulin resis- 85. Cai, D., et al. 2004. IKKβ/NF-κB activation causes 95. Castrillo, A., and Tontonoz, P. 2004. Nuclear recep-
tance. Proc. Natl. Acad. Sci. U. S. A. 100:7265–7270. severe muscle wasting in mice. Cell. 119:285–298. tors in macrophage biology: at the crossroads of
75. Weisberg, S.P., et al. 2003. Obesity is associated with 86. Dominguez, H., et al. 2005. Metabolic and vascu- lipid metabolism and inflammation. Annu. Rev.
macrophage accumulation in adipose tissue. J. Clin. lar effects of tumor necrosis factor-alpha blockade Cell Dev. Biol. 20:455–480.

The Journal of Clinical Investigation      http://www.jci.org      Volume 116      Number 7      July 2006 1801

You might also like